• Non ci sono risultati.

Nuclear factor kappa B in patients with a history of unstable angina: case re-opened

N/A
N/A
Protected

Academic year: 2021

Condividi "Nuclear factor kappa B in patients with a history of unstable angina: case re-opened"

Copied!
9
0
0

Testo completo

(1)

https://doi.org/10.1007/s11739-018-1885-z IM - ORIGINAL

Nuclear factor kappa B in patients with a history of unstable angina:

case re‑opened

Chiara Mozzini1  · Ulisse Garbin1 · Chiara Stranieri1 · Giulia Salandini1 · Giancarlo Pesce2 ·

Anna Maria Fratta Pasini1 · Luciano Cominacini1

Received: 21 August 2017 / Accepted: 26 May 2018 © SIMI 2018

Abstract

This study aims at assessing NF-kB activity in unstable angina (UA) patients free of symptoms after a 1 year follow-up (1YFU). Plasma oxidized low-density lipoproteins (oxLDL), circulating NF-kB, Interleukin 6 (IL-6) and Interleukin 1β (IL-1β), high-sensitivity C-reactive protein (hs-CRP), as markers of oxidative stress and inflammation and plasma double-stranded DNA (ds-DNA), as marker of Neutrophil Extracellular Traps (NETs), were measured in 23 of the previously enrolled 27 UA patients. These measurements were compared to the UA data at baseline, and then compared to the data derived from the stable angina (SA) and controls (C) enrolled in our previous study (we demonstrated that UA had higher levels of NF-kB compared to SA and C). After a 1YFU, UA patients show a significant decrease in NF-kB, IL-6, hs-CRP, oxLDL, and ds-DNA plasma levels (p < 0.001) and in IL-1β and White Blood Cells (WBC) (p < 0.005), without differences in lipid and glucose assessment. If compared to SA and C, UA after a 1YFU have higher levels of NF-kB, IL-6, ds-DNA, WBC, and oxLDL compared to C (p < 0.001), but only IL-6 is higher than SA (p < 0.001). No differences are found in lipid and glucose assessment. After a 1YFU, patients with a history of UA improve their oxidative and inflammatory status, such as the levels of circulating ds-DNA, without achieving the status of C. They become comparable to SA subjects. This study provides new insight on the multiple and apparently contradictory facets of NF-kB in UA and on its possible role as media-tor in NETs’ formation.

Keywords Unstable and stable angina · Nuclear factor kappa B · Inflammation · Double-stranded DNA · NETosis

Introduction

Coronary artery disease is a common complex atheroscle-rotic pathology associated with substantial morbidity and mortality [1]. Unstable angina (UA) is defined as myocar-dial ischemia at rest or minimal exertion in the absence of cardiomyocyte necrosis [2]. Compared with non-ST-ele-vation myocardial infarction patients, individuals with UA do not experience myocardial necrosis, have a substantially lower risk of death, and appear to derive less benefit from

intensified anti-platelet therapy as well as early invasive strategy [3, 4]. UA prevalence is expected to be 10% [5] among unselected patients admitted with acute chest pain to the emergency department (ED).

Nuclear factor kappa B (NF-kB) is the central regulator of innate and adaptive response with hundreds of target genes, some with pro-inflammatory effects, and some promoting cell survival [6]. NF-kB intervenes in the transcription of a large number of inflammatory genes coding for cytokines, chemokines, and adhesion molecules [7]. NF-kB can be acti-vated via the canonical and the non-canonical pathway [7]. NF-kB is normally held in the cytoplasm in complex with the inhibitor-kBα (IkBα). Canonical activation of NF-kB involves phosphorylation of IkBα and its proteasome deg-radation when inflammation occurs [8]. This pathway is mainly activated in response to pro-inflammatory stimuli. Non-canonical NF-kB signalling is important for the devel-opment and maintenance of primary and secondary lym-phoid organs, and adaptive immune responses [7, 8].

* Chiara Mozzini [email protected]

1 Department of Medicine, Section of Internal Medicine, University of Verona, Piazzale L.A. Scuro, 10 - 37134 Verona, Italy

2 Department of Diagnostic and Public Health, Unit of Epidemiology and Medical Statistics, University of Verona, Piazzale L.A. Scuro, 10 - 37134 Verona, Italy

(2)

A key role for NF-kB is essential in the pathophysiol-ogy of myocardial re-perfusion injury, ischemic precon-ditioning, and UA [9].

Nevertheless, the protective role for NF-kB during pathological remodelling of the heart is a source of con-troversy [9]. This is due to the evidence that NF-kB also regulates many different anti-apoptotic factors, such as cel-lular inhibitors of apoptosis, caspase inhibitors, and Bcl-2 family members [10].

Cell-free DNA (cf-DNA) is present in small amounts in plasma of healthy subjects [11], but it has been reported to be elevated in various clinical disorders [12]. It has been correlated with the degree of tissue damage, originating from necrosis and apoptosis of blood and tissue cells [12]. In particular, cf-DNA has been found to be elevated in patients with acute coronary syndrome in different studies [13, 14] with a prognostic potential [15].

Part of cf-DNA is double-stranded DNA (ds-DNA). Ds-DNA is a marker of the peculiar process through which neutrophils and other cell types expel ds-DNA [16, 17]. A fascinating novel explanation of how DNA can actively be released under inflammatory conditions has recently been discovered [16, 17]. It is an evolutionary and highly conserved first-line defence mechanism that allows neu-trophils to expel their DNA, forming a meshwork of chro-matin and proteins termed neutrophil extracellular traps (NETs) [16, 17]. NETs are the results of a peculiar form of cell death that is morphologically characterized by the loss of intracellular membranes before the integrity of the plasma membrane can be lost. The death of neutrophils with NETs formation is called NETosis. The main function of NETs is trapping and killing pathogens. Nevertheless, recent studies suggest NETs interference in other diseases (venous thromboembolism, cancer, autoimmune diseases [18–22]) and in atherosclerosis progression [23–26], as recently reviewed [27].

We have previously demonstrated [28] that UA patients have higher levels of NF-kB compared to stable angina (SA) patients, and both have higher levels compared to coronary artery disease-free controls (C). The activation of NF-kB in circulating cells of UA patients is, at least in part, induced by oxidized low-density lipoproteins (oxLDL).

The purpose of the present study is to assess NF-kB activity and related-circulating molecules in the same UA patients now free of symptoms after a 1 year follow-up (1YFU).

This study aims also to provide new insight on the mul-tiple and apparently contradictory facets of NF-kB in UA, discussing its deleterious but also its less known survival-promoting effects. Moreover, a new field of research is pro-posed concerning the possible role of NF-kB as mediator in NETs formation.

Materials and methods

Ethical considerations

The study was conducted in accordance with the ethical stand-ards laid down in the Helsinki Declaration of 1975 and its late amendments. All participants provided written consent prior to commencing the study and the local ethical committee (Uni-versity of Verona-Azienda Ospedaliera Universitaria Integrata Verona) approved the study.

Recruitment of participants

The study population and the measurement methods of NF-kB activity and related-circulating molecules have been previously described [28]. In addition, exclusion criteria were maintained as explained in [28].

As described [28], UA were patients with at least two epi-sodes of rest anginal pain or one episode lasting more than 20 min in the previous 48 h preferably, but not necessarily, associated with electrocardiographic modifications (T-wave inversion, ST-segment depression, and transient ST-segment elevation) and a normal value of I-troponin on admission and during the first 24 h.

All 27 previously enrolled UA patients were recalled after 1 year and a blood sample collection was proposed.

Blood sample collection, peripheral blood mononuclear cells (PBMC) isolation, oxidized low-density lipoproteins (oxLDL) plasma levels, circulating NF-kB, high-sensitivity C-Reactive Protein (hs-CRP), lipid assessment, and white blood cells (WBC) were evaluated as previously described [28]. Circulating Interleukin (IL)-6 and 1-β were tested accord-ing to the methods described in our previous study [29].

Fluorescent assay Quant-iT™ PicoGreen® ds-DNA

Rea-gent and Kits (Invitrogen) [30] has been used to measure ds-DNA in serum of UA, SA, C, and UA 1YFU patients. Statistical analysis

Data were summarized as mean ± standard deviation or median (first quartile; third quartile) for normally and non-normally distributed variables, respectively. Differences values were tested using a paired-sample Student’s t test or Wilcoxon matched-pair signed-rank test, accordingly to the type of distri-bution. Statistical analyses were performed with STATA 14.1 and a 0.05 significance level was adopted.

Results

Twenty-three of the twenty-seven previously enrolled UA patients accepted the 1YFU-recall.

(3)

Two patients (males) were excluded for recent coronary artery by-pass grafting, one patient (male) was excluded for malignancy onset, and one patient (male) denied his consent to the 1YFU evaluation. Then, the study setting was com-posed of 23 UA patients after 1YFU (UA1YFU): 3 females and 20 males now free of symptoms related to angina.

Baseline clinical characteristics of the patients are listed in Table 1 in [28] and [29].

Nevertheless, a summary table modified from this has been re-created: Table 1. Drug therapy was now similar in the UA1YFU group: acetylsalicylic acid, angiotensin-con-verting enzyme inhibitor, β-blocker, and statin.

Table 2 depicts the distribution of concentrations of activated circulating NF-kB in PBMC, serum ds-DNA, circulating cytokines, OxLDL, lipid and glucose assess-ment, and hs-CRP and WBC levels of UA patients at base-line and after a 1YFU. No significant differences in plate-lets count or in Mean Platelet Volume (MPV) are found (210.000 ± 5000 109/L and 91 fL for UA at baseline versus

200.000 ± 4000 109/L and 93 fL after 1YFU, p 0.03).

NF-kB, IL-6, hs-CRP, oxLDL, and ds-DNA levels are significantly lower in UA patients after 1YFU, compared to the year before (p < 0.001).

Table 1 Baseline clinical characteristics of the groups of patients (modified from Table 1 of [28])

Data are expressed in percent

C controls, UA unstable angina patients, UA1YFU unstable angina patients after a 1 year follow-up, ACE-I angiotensin-converting enzyme inhibitors, ACS acute coronary syndrome, PCACE-I percutaneous coronary

intervention, NS not significantly different, / not re-calculated

C (n = 27) SA (n = 29) UA (n = 27) p value UA1YFU (n = 23) p value

Age (years) 66 ± 11 61 ± 9 61 ± 9 NS 62 ± 6 NS Family history 37% 66% 38% NS 37% / Smoke 18% 28% 46% NS 0% none < 0.01 Hypercholesterolemia 41% 79% 69% < 0.05 68% / Hypertension 48% 79% 61% NS 60% / Diabetes 7% 14% 15% NS 15% / ACE-I 63% 59% 35% NS 100% < 0.01 Statins 11% 24% 11% NS 100% < 0.01 Aspirin 30% 76% 50% < 0.05 100% < 0.01

Previous ACS None 65% 31% < 0.01 30% /

Previous PCI None 45% 8% < 0.01 7% /

Table 2 Laboratory data of UA patients at baseline and after a 1YFU

Normally distributed continuous variables are expressed as mean ± standard deviation, while non-normally distributed variables are presented as median and interquartile range

UA unstable angina patients, UA1YFU unstable angina patients after a 1 year follow-up, NF-kB nuclear

fac-tor kappa B, IL-6 Interleukin 6, IL-1β Interleukin 1β, hs-CRP high-sensitivity C-Reactive protein, oxLDL oxidized low-density lipoprotein, LDL low-density lipoprotein, HDL high-density lipoprotein, WBC white blood cells, ds-DNA double-stranded DNA

UA baseline (n = 23) LIAlYFU (n = 23) p value

NF-kB ng/μg cell protein 1.52 (1.54; 1.72) 0.94 (0.35; 1.2) < 0.001 IL-6 pg/mL 6.93 ± 3.43 3.03 ± 1.91 < 0.001 IL-1β pg/mL 0.79 (0.17; 1.37) 0.15 (0.01; 0.28) 0.016 hs-CRP mg/dL 0.98 (0.08; 1.21) 0.42 (0.26; 0.53) < 0.001 oxLDL μg/mL 38.7 ± 4.39 23.1 ± 3.54 < 0.001 LDL mg/dL 140 ± 26 128 ± 27 0.264 HDL mg/dL 44.3 ± 3.6 48.3 ± 11.2 0.145 Cholesterol mg/dL 215 ± 39 202 ± 33 0.302 Triglycerides mg/dL 149 ± 54 160 ± 50 0.819 Glucose mg/dL 101 ± 15 103 ± 46 0.361 WBC count 109/L 9.53 ± 1.8 7.30 ± 1 0.002 ds-DNA ng/mL 23 ± 1 14 ± 1 < 0.001

(4)

IL-1β and WBC levels are significantly lower in UA patients after 1YFU compared to the year before (p < 0.005). No significant differences are found in routine lipid and glu-cose assessment.

Figures 1, 2 show, respectively, the levels of NF-kB (expressed in ng/μg cell protein) and ds-DNA (expressed in ng/mL) in UA patients at baseline and after a 1YFU, com-pared with SA patients and C.

NF-kB (Fig. 1) and ds-DNA (Fig. 2) levels in UA1YFU are significantly lower compared to UA baseline (p < 0.001) and not significantly different compared to SA.

All data (for UA baseline, UA1YFU and SA) are signifi-cantly higher compared to C (p < 0.001).

Figure 3 shows blood concentrations of IL-6 and IL-1β in UA at baseline, UA1YFU, SA, and C patients. As previ-ously reported [29], IL-6 and IL-1β levels were significantly higher (p < 0.001) in UA compared to SA and C. UA1YFU patients show lower levels of IL-6 (p < 0.001) compared to UA at baseline, but significantly higher levels compared to SA and C (p < 0.001). UA1YFU patients show lower levels of IL-1β (p < 0.001) compared to UA at baseline, but no significant differences were found if compared to SA and C.

A significant difference (p < 0.002) is found in WBC in UA after a 1YFU (7.30 ± 1 109/L) compared to UA at

base-line (9.53 ± 1.8 109/L). UA1YFU patients have higher WBC

levels compared both to SA and C (respectively, 6 ± 0.2 and 6 ± 0.1 109/L; p < 0.005).

A significant difference (p < 0.001) is found in hs-CRP in UA after a 1YFU, 0.42 (0.26; 0.53) mg/dL, compared to UA at baseline, 0.98 (0.08; 1.21) mg/dL. No significant dif-ferences in hs-CRP levels are found for UA1YFU patients compared to SA and C, respectively, 0.50 (0.08; 1) and 0.43 (0.08; 0.8) mg/dL.

A significant difference (p < 0.001) is found in oxLDL in UA after a 1YFU (23.1 ± 3.54 μg/mL) compared to UA at

baseline (38.7 ± 4.39 μg/mL). UA1YFU patients have higher oxLDL levels compared to C (12.9 ± 4 μg/mL; p < 0.001), but no significant differences are found compared to SA (23.3 ± 4.4 μg/mL).

Discussion

The main finding of this work is that, after a 1YFU, patients with a history of UA improve their inflammatory status, but without achieving the status of C, and becoming comparable to SA subjects.

The focus of this manuscript is to investigate the role of NF-kB in UA patients in their follow-up.

NF-kB has been largely investigated in cardiovascular diseases, in particular in ischemic heart disease [9, 10, 28,

29].

NF-kB is generally well known to worsen cardiac remod-elling by activating pro-inflammatory pathways to mediate cardiac hypertrophy and maladaptive remodelling [31, 32].

Fig. 1 Concentrations of activated circulating Nuclear Factor kappa B (NF-kB) in unstable angina (UA) patients at baseline and after a 1YFU (UA1YFU), compared to stable angina (SA) patients and con-trol subjects (C). The NF-kB was extracted from peripheral blood mononuclear cells (PBMC) derived from the patients. *p < 0.001 vs C; ƚp < 0.001 vs SA; §p < 0.001 vs UA1YFU

Fig. 2 Serum ds-DNA of unstable angina (UA) patients at base-line and after a 1YFU (UA1YFU), compared to stable angina (SA) patients and control subjects (C). *p < 0.001 vs C; ƚp < 0,001 vs SA; §p < 0.001 vs UA1YFU

Fig. 3 Plasma concentrations of IL-6 and IL-1β in unstable angina (UA) patients at baseline and after a 1YFU (UA1YFU), com-pared to stable angina (SA) patients and control subjects (C). IL-6: *p < 0.001 vs C; ƚp < 0.001 vs SA. IL-1β: °p < 0.001 vs C; #p < 0.001 vs UA1YFU and SA

(5)

However, the intriguing point concerns the multi-faceted role of NF-kB, in particular its protective role as emerged in different studies mentioned below. This fact opens a debate about the deleterious and protective role of this transcription factor, as recently reviewed [33].

In fact, NF-kB is able to induce the expression of several survival proteins including c-IAP1 and 2, and TRAF-1 and 2 [34–36]. Moreover, NF-kB exerts its protective effects by up-regulating the expression of several genes such as Bcl-2 family members and caspase inhibitors [37].

NF-kB protective action has been shown to be related to the cross-talk with heat shock proteins, normally implicated in the protection against apoptosis [38–40]. The involvement of NF-kB in ischemia re-perfusion injury and protective pathways has been observed [41–44]. In this study, NF-kB levels are significantly lower in UA patients after a 1YFU compared to the year before, and after a 1YFU, they are comparable to SA. The activation of NF-kB in circulating cells of UA patients is, at least in part, induced by oxLDL, as previously demonstrated [28]. The NF-kB persistent activa-tion, also is lower compared to the acute event (with lower levels of oxLDL), can be explained with the double role of this transcription factor, regulating its action according to the different situations.

Inflammation has a well-established role in coronary artery disease [45, 46]. As reported in the previous authors’ study [28], the total WBC count and CRP levels are signifi-cantly higher in UA patients compared to SA and C. After a 1YFU, UA patients have significantly lower levels of these markers compared to their baseline. This fact is important, because it opens a debate about the usefulness of CRP as a precise follow-up marker. In a prospective multicentre study [47], CRP was measured at admission, at hospital discharge and 1 month later in consecutive patients hospitalized for acute coronary syndrome. This study aimed to determine whether there was a clinical prognostic utility for measuring CRP following an acute coronary episode. In conclusion, the study did not support the clinical use of CRP because of the lack of substantial predictive ability (death, myocardial infarction, or UA) of this marker. Nevertheless, CRP is con-sidered the principal marker able to predict short- and long-term outcome in patients with acute coronary syndrome, with several studies supporting this notion [48–50].

Then, the discussion could be driven toward a particular field of research with the aim of including a proposal of the possible role of NF-kB as mediator in NETs’ formation.

According to the results of this study, patients with UA at baseline have significantly higher levels of circulating ds-DNA compared to SA and C. After 1YFU, the UA patients became comparable to SA.

To our knowledge, this is the first study that after a 1YFU compares ds-DNA in UA at baseline to SA and C. Sus-tained levels of ds-DNA in UA1YFU and SA patients might

indicate that also even without symptoms, these groups of patients are not comparable to healthy subjects. This fact could be revelatory of ongoing tissue damage, but also of the activation of neutrophils in forming NETs. Moreover, UA1YFU patients have higher WBC levels compared both to SA and C. The increased number and the stimulation of neutrophils (represented by ds-DNA) might be driven by NF-kB. In fact, a recent study [51] has shown that NF-kB is involved in the generation of NETs. The authors demonstrate that acetylsalicylic acid (ASA) and NF-kB inhibitors (using two structurally specific inhibitor of IkBα phosphorylation) have an inhibitory effect on NETs formation in vitro. The effects of ASA are not only related to cyclo-oxygenase acety-lation, but also include inhibition of NF-kB activation [52]. A previous study [53] shows that the stimulation of neutro-phils induces both the nuclear accumulation of NF-kB/Rel proteins and the concomitant degradation of IkBα.

On the basis of these studies, Lapponi et al. [51] conclude that this major inflammatory factor is involved in the inflam-matory response mediated by NETs.

Ds-DNA has cytotoxic and pro-thrombotic effects [54], creating a link between inflammation and coagulation. The study by Borissoff [23] clarifies the relationships between extracellular DNA formation, coronary atherosclerosis, and the presence of a pro-thrombotic state. The study reveals that markers of NETosis (ds-DNA, nucleosomes, citrul-linated histone H4, and MPO-DNA complexes) are inde-pendently associated with the severity of coronary artery disease, a pro-thrombotic state, and also the occurrence of major adverse cardiac events. The study suggests that NETs formation might contribute to atherosclerosis progression.

The previous histological studies have shown the pres-ence of NETs in the luminal portion of mouse and human atherosclerotic lesions [24, 25]. In a very recent study [26], coronary thrombarterectomies derived from patients with ST-elevation acute coronary syndrome (undergoing primary percutaneous coronary intervention) were analyzed. In the culprit lesion site, NETs burden positively correlate with infarct size and negatively with ST-segment resolution. In fact, nucleosomes, ds-DNA, neutrophil elastase, myeloper-oxidase, all markers of NETosis are found to be increased in the culprit lesion site [26].

Inflammation contributes to all phases of atherosclerosis [55]. Interleukins, in particular, IL-6 and IL-1β, are critical mediators of the systemic inflammatory response [56, 57].

Secretion of cytokines by inflammatory cells is a major driver of pathogenesis in UA [58]. As previously reported by the authors [29], UA patients exhibit greatly enhanced plasma concentrations of IL-6 and IL-1β compared to SA and C. As discussed [29], this fact might be related to the effect of circulating oxLDL.

Now, after 1YFU, UA patients show lower levels of IL-6 compared to UA at baseline, but significantly higher levels

(6)

compared to SA and C. UA1YFU patients show lower levels of IL-1β compared to UA at baseline, but no significant dif-ferences are found if compared to SA and C. Thus, the trend of these molecules is quite different.

It is well known that interleukin-1 (IL-1) plays a particu-larly prominent role in atherothrombosis [59]. In addition, a complex of intracellular proteins, known as the nucleo-tide-binding leucine-rich repeat-containing pyrin receptor 3 (NLRP3) inflammasome, activates caspase-1 or IL-1β converting enzyme, the protease that produces active IL-1β from its inactive precursor [60–63]. Several exogenous “dan-ger signals” trig“dan-ger the inflammasome including crystalline compounds. It has been shown that the NLRP3 inflamma-some is critical for production of active IL-1β responses not only by bacteria, crystalline uric acid, and crystalline pyrophosphate, but also to cholesterol crystals and mini-mally modified LDL cholesterol [64].

These facts were the basis of the well-known CANTOS study [65] with Canakinumab.

This fact leads to some considerations, trying to give a link between these cytokines, NETs, and NF-kB regulation.

In a mouse model of atherosclerosis [66], cholesterol crystals act both as priming and danger signals for IL-1β production. Cholesterol crystals trigger neutrophils to release NETs.

In addition, IL-6 is a potent NETs inducer, as previously demonstrated [67]. Nevertheless, this cytokine is principally known for a precise role in atherosclerosis [68]. Large quan-tities of IL-6 were found in human atherosclerotic plaques [69]. In particular, IL-6 can promote the occurrence of ath-erosclerosis development and plaque rupture [68, 69].

Different studies [70–72] show that serum IL-6 of acute myocardial infarction patients is significantly higher com-pared to UA patients. The levels in UA are significantly higher compared to SA. These results agree with the result of the current study. The potential causal role of IL-6 in atherothrombosis has been suggested by its selective expres-sion in macrophages in murine and human atheroma [69,

73, 74]. IL-6 is highly up-regulated at the site of the cor-onary occlusion. It can be produced by cardiac myocytes under condition of local hypoxia in the viable border zone of re-perfused infarction. [75]. IL-6 is considered a predic-tor of high-risk coronary anatomy, as defined by coronary computed tomography angiography [76]. At least, circulat-ing IL-6 has been shown to be associated with the thin-cap fibroatheroma, that is the lesion with the highest potential for plaque rupture [77, 78]. All these data suggest that IL-6 levels might correlate with the instability of the atheroscle-rotic plaque. In fact, IL-6 has a stimulatory effect on smooth muscle cells proliferation [77].

Il-6 is regulated by NF-kB [79, 80]. The hallmark of vas-cular NF-kB activation is the production of IL-6, whose local role in vascular inflammation has been reviewed [79].The

ultimate consequence of NF-kB signalling is the activation of inflammatory genes including adhesion molecules and chemo-taxins. However, clinically, the hallmark of vascular NF-kB activation is the production of IL-6 [79].The same trend in UA at baseline, UA1YFU, SA for IL-6, and NF-kB may reflect this fact. In particular, the higher levels in UA1YFU compared to SA might correlate with the plaque instability that led to the UA condition the year before.

All the markers considered in this work are affected by therapy, of course. There is a large knowledge about this fact [26, 29, 45, 50–52, 65]. The small sample of patients in the current study does not allow the exact contribution of each drug in the influence of the final condition. However, the find-ing that despite complete therapy (accordfind-ing to the current guidelines for UA), UA1YFU patients do not reach the condi-tion of C; this fact itself means that the pathology itself makes the difference.

Study limitations

The main limitation of this study is the lack of complete collec-tion of NETosis markers other than ds-DNA. This fact makes the authors’ considerations partially elusive for the moment.

Moreover, several biological and methodological hurdles have been identified in cf-DNA testing, as reviewed [81] (dif-ferent testing methods, great variability of the levels in the healthy population, etc.). Nevertheless, up to now, no precise data are available about ds-DNA. It is reasonable that similar considerations could also be done for these DNA fragments.

The absence of a follow-up sampling in the SA and C is a further consistent limitation of the study.

However, the strength of the study is underlining the new overview of the role of NF-kB, as a protective factor con-nected with NETosis in UA. These notions must be elevated to a new degree when considering the enormously compli-cated interacting networks that explain the complex and not fully investigated mechanisms that link immunity, inflamma-tion, and cardiovascular diseases. There are very few data in the literature about the hypothesis that NF-kB may act as the mediator of NETs’ formation. A very recent paper, in a different context (Dermatology and Wound healing), analyzes this fact. NETs’ scaffold recognized by Toll-Like Receptor 9 (TLR 9) is able to activate the NF-kB pathway. NETs’ stimulation rapidly induces a dose dependent NF-kB activation and such signalling pathway modulates keratino-cytes proliferation [82].

Conclusions

After a 1YFU, patients with a history of UA improve their inflammatory status, but without achieving the status of C, and becoming comparable to SA subjects.

(7)

In conclusion, the persistent activation of NF-kB in these patients might also be considered a conceivable solution to maintain an innate immunity response, as NETosis is. NF-kB activation and NETs formation are, therefore, similar to a double-edged sword, acting not only as an effective first-line defence mechanism, but also leading to organ failure and death if the process is uncontrolled.

Author contributions CM and LC conceived the study; GP statistically analyzed the data; AF and UG revised the data and the manuscript; GS and CS performed the experiments; CM wrote the manuscript. Compliance with ethical standards

Conflict of interest The Authors declare that they have no conflict of interest.

Statement of human and animal rights The study was conducted in accordance with the ethical standards laid down in the Helsinki Dec-laration of 1975 and its late amendments.

Informed consent Informed consent was obtained from all individual participants included in the study.

References

1. Mathers CD, Loncar D (2006) Projection of global mortality and burden of disease from 2002 to 2030. PLoS Med 3:2011–2030. https ://doi.org/10.1371/journ al.pmed.00304 42

2. Task Force for the Management of Acute Coronary Syndromes in Patients Presenting without Persistent ST-Segment Elevation of the European Society of Cardiology (ESC) (2016) 2015 ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation. Eur Heart J 37:267–315. https ://doi.org/10.1093/eurhe artj/ehv32 0 3. Thygesen K, Mair J, Giannitsis E, Mueller C, Lindahl B,

Blanken-berg S, Huber K, Plebani M, Biasucci LM, Tubaro M, Collinson P, Venge P, Hasin Y, Galvani M, Koenig W, Hamm C, Alpert JS, Katus H, Jaffe AS (2012) How to use high-sensitivity cardiac troponins in acute cardiac care. Eur Heart J 33:2252–2257. https ://doi.org/10.1093/eurhe artj/ehs15 4

4. Braunwald E, Morrow DA (2013) Unstable angina: is it time for a requiem? Circulation 127:2452–2457. https ://doi.org/10.1161/ CIRCU LATIO NAHA.113.00125 8

5. Mockel M, Searle J, Hamm C, Slagman A, Blankenberg S, Huber K, Katus H, Liebetrau C, Muller C, Muller R, Peitsmeyer P, von Recum J, Tajsic M, Vollert JO, Giannitsis E (2015) Early discharge using single cardiac troponin and copeptin testing in patients with suspected acute coronary syndrome (ACS): a rand-omized, controlled clinical process study. Eur Heart J 36:369–376. https ://doi.org/10.1093/eurhe artj/ehu17 8

6. Kumar A, Takada Y, Borick AM, Aggarwal BB (2004) Nuclear factor kappa B: its role in health and disease. J Mol Med 82:434– 448. https ://doi.org/10.1007/s0010 9-004-0555-y

7. Mitchell S, Vargas J, Hoffmann A (2016) Signaling via the NF-kB system. Syst Biol Med 8:227–241. https ://doi.org/10.1002/ wsbm.1331

8. Hayden MS, Ghosh S (2008) Shared principles in NF-kappa B signaling. Cell 132:344–362. https ://doi.org/10.1016/j. cell.2008.01.020

9. Gordon JW, Shaw JA, Kirshenbaum LA (2011) Multiple fac-ets of NF-kB in the heart: to be or not to NF-kB. Circ Res 108:1122–1132. https ://doi.org/10.1161/CIRCR ESAHA .110.22692 8

10. Karin M, Lin A (2002) NF-kB at the crossroads of life and death. Nat Immun 3:221–227. https ://doi.org/10.1038/ni030 2-221 11. Suzuki N, Kamataki A, Yamaki J, Homma Y (2008)

Characteriza-tion of circulating DNA in healthy human plasma. Clin Chim Acta 387:55–59. https ://doi.org/10.1016/j.cca.2007.09.001

12. Swarup V, Rajeswari A (2007) Circulating (cell-free) nucleic acids—a promising non-invasive tool for early detection of several human diseases. FEBS Lett 581:795–799. https ://doi. org/10.1016/j.febsl et.2007.01.051

13. Cui M, Fan M, Jing R, Wang H, Qin J, Sheng H, Wang Y, Wu X, Zhang L, Zhu J, Ju S (2013) Cell-free circulating DNA: a new bio-marker for the acute coronary syndrome. Cardiology 124(2):76– 84. https ://doi.org/10.1159/00034 5855

14. Chang CP, Chia RH, Wu TL, Tsao KC, Sun CF, Wu JT (2003) Elevated cell-free serum DNA detected in patients with myocar-dial infarction. Clin Chim Acta 327(1–2):95–101

15. Destouni A, Vrettou C, Antonatos D, Chouliaras G, Traeger-Synodinos J, Patsilinakos S, Kitsiou-Tzeli S, Tsigas D, Kanava-kis E (2009) Cell-free DNA levels in acute myocardial infarction patients during hospitalization. Acta Cardiol 64(1):51–57. https ://doi.org/10.2143/AC.64.1.20343 62

16. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y, Zychlinsky A (2004) Neutrophil extra-cellular traps kill bacteria. Science 303(5663):1532–1535. https ://doi.org/10.1126/scien ce.10923 85

17. Fuchs TA, Abed U, Goosmann C, Hurwitz R, Schulze I, Wahn V, Weinrauch Y, Brinkmann V, Zychlinsky A (2007) Novel cell death program leads to neutrophil extracellular traps. J Cell Biol 176(2):231–241. https ://doi.org/10.1083/jcb.20060 6027 18. Marcos V, Zhou Z, Yildirim AO, Bohla A, Hector A, Vitkov L,

Wiedenbauer E-M, Krautgartner WD, Stoiber W, Belohradsky BH (2010) CXCR2 mediates NADPH oxidase-independent neutrophil extracellular trap formation in cystic fibrosis airway inflammation. Nat Med 16:1018–1023. https ://doi.org/10.1038/nm.2209 19. Sangaletti S, Tripodo C, Chiodoni C, Guarnotta C, Cappetti B,

Casalini P, Piconese S, Parenza M, Guiducci C, Vitali C (2012) Neutrophil extracellular traps mediate transfer of cytoplasmic neu-trophil antigens to myeloid dendritic cells toward ANCA induc-tion and associated autoimmunity. Blood 120:3007–3018. https ://doi.org/10.1182/blood -2012-03-41615 6

20. Brinkmann V, Zychlinsky A (2012) Neutrophil extracellular traps: is immunity the second function of chromatin? J Cell Biol 198:773–783. https ://doi.org/10.1083/jcb.20120 3170

21. Demers M, Krause DS, Schatzberg D, Martinod K, Voorhees JR, Fuchs TA, Scadden DT, Wagner DD (2012) Cancers predis-pose neutrophils to release extracellular DNA traps that contrib-ute to cancer-associated thrombosis. Proc Natl Acad Sci USA 109:13076–13081. https ://doi.org/10.1073/pnas.12004 19109 22. Demers M, Wagner DD (2014) NETosis: a new factor in tumour

progression and cancer-associated thrombosis. Semin Thromb Haemost 40(3):277–283. https ://doi.org/10.1055/s-0034-13707 65 23. Borissoff JI, Joosen IA, Versteylen MO, Brill A, Fuchs TA,

Savchenko AS, Gallant M, Martinod K, Ten Cate H, Hofstra L, Crijns HJ, Wagner DD, Kietselaer BL (2013) Elevated levels of circulating DNA and chromatin are independently associated with severe coronary atherosclerosis and a prothrombotic state. Arterioscler Thromb Vasc Biol 33(8):2032–2040. https ://doi. org/10.1161/ATVBA HA.113.30162 7

24. Megens RT, Vijayan S, Lievens D, Döring Y, van Zandvoort MA, Grommes J, Weber C, Soehnlein O (2012) Presence of luminal neutrophil extracellular traps in atherosclerosis. Thromb Haemost 107(3):597–598. https ://doi.org/10.1160/TH11-09-0650

(8)

25. De Boer OJ, Li X, Teeling P, Mackaay C, Ploegmakers HJ, van der Loos CM, Daemen MJ, de Winter RJ, van der Wal AC (2013) Neutrophils, neutrophil extracellular traps and interleukin-17 associate with the organisation of thrombi in acute myocar-dial infarction. Thromb Haemost 109(2):290–297. https ://doi. org/10.1160/TH12-06-0425

26. Mangold A, Alias S, Scherz T, Hofbauer T, Jakowitsch J, Panzen-böck A, Simon D, Laimer D, Bangert C, Kammerlander A, Mascherbauer J, Winter MP, Distelmaier K, Adlbrecht C, Pre-issner KT, Lang IM (2015) Coronary neutrophil extracellular trap burden and deoxyribonuclease activity in ST-elevation acute coronary syndrome are predictors of ST-segment resolution and infarct size. Circ Res 116(7):1182–1192. https ://doi.org/10.1161/ CIRCR ESAHA .116.30494 4

27. Mozzini C, Garbin U, Fratta Pasini AM, Cominacini L (2016) An exploratory look at NETosis in atherosclerosis. Intern Emerg Med. https ://doi.org/10.1007/s1173 9-016-1543-2

28. Cominacini L, Anselmi M, Garbin U, Fratta Pasini A, Stranieri C, Fusaro M, Nava C, Agostoni P, Keta D, Zardini P, Sawamura T, Lo Cascio V (2005) Enhanced plasma levels of oxidized low-density lipoprotein increase circulating nuclear factor-kappa B activation in patients with unstable angina. J Am Coll Cardiol 46:799–806. https ://doi.org/10.1016/j.jacc.2005.05.063

29. Fratta Pasini AM, Anselmi M, Garbin U, Franchi E, Stranieri C, Nava MC, Boccioletti V, Vassanelli L, Cominacini L (2007) Enhanced levels of oxidized low density lipoprotein prime mono-cytes to cytokine overproduction via upregulation of CD14 and Toll-like receptor 4 in unstable angina. Arterioscler Thromb Vasc Biol 27:1991–1997. https ://doi.org/10.1161/ATVBA HA.107.14269 5

30. Lindorfer MA, Schuman TA, Craig ML, Martin EN, Taylor RP (2001) A bispecific dsDNA monoclonal antibody construct for clearance of anti-dsDNA IgG in systemic lupus erythematosus. J Immunol Methods 248(1–2):125–138

31. Li Y, Ha T, Gao X, Kelley J, Williams DI, Browder IW, Kao RI, Li C (2004) NF-kB activation is required for the development of cardiac hypertrophy in vivo. Am J Physiol 287:H1712–H1721. https ://doi.org/10.1152/ajphe art.00124 .2004

32. Frantz S, Hu K, Bayer B, Gerondakis S, Strottmann J, Adamek A, Ertl G, Bauersachs J (2006) Absence of NF-kB subunit p50 improves heart failure after myocardial infarction. FASEB J 20:1918–1920. https ://doi.org/10.1096/fj.05-5133fj e

33. Dhingra R, Shaw JA, Aviv Y, Kirshenbaum LA (2010) Dichoto-mous actions of NF-kappaB signaling pathways in heart. J Car-diovasc Transl Res 4:344–354. https ://doi.org/10.1007/s1226 5-010-9195-5

34. You M, Ku PT, Hrdlikova R, Bose HR (1997) Ch-IAP-1 member of the inhibitor of apoptosis protein family is a mediator of the antiapoptotic activity of the v-Rel oncoprotection. Mol Cell Biol 17:7328–7341

35. Chu ZL, Mc Kinsey TA, Liu L, Gentry JJ, Malim MH, Ballard DW (1997) Suppression of tumour necrosis factor induced death by inhibitor of apoptosis c-IAP2 is under NF-kB control. Proc Natl Acad Sci 94:10057–10062

36. Wang CY, Mayo MW, Korneluk RG, Goeddel DV, Baldwin AS (1998) NF-kB antiapoptosis: induction of TRAF1 and TRAF2 and cIAP1 and c-IAP2 to suppress caspase-8 activation. Science 281:1680–1683

37. Papa S, Bubici C, Zazzeroni F, Pham CG, Kuntzen C, Knabb JR, Dean K, Franzoso G (2006) The NF- kB- mediated control of JNK cascade in the antagonism of programmed cell death in health and disease. Cell Death Differ 13:712–729. https ://doi.org/10.1038/ sj.cdd.44018 65

38. Beere HM (2004) The stress of dying: the role of heat shock pro-teins in the regulation of apoptosis. J Cell Sci 117:2641–2651. https ://doi.org/10.1242/jcs.01284

39. Beere HM (2005) Death versus survival: functional interac-tion between the apoptotic and stress-inducible heat shock protein pathways. J Clin Investig 115:2633–2639. https ://doi. org/10.1172/JCI26 471

40. Valen G, Hansson GK, Dumitrescu A, Vaage J (2000) Unstable angina activates myocardial heat shock protein 72, endothelial nitric oxide synthase and transcription factor NF kappa B and AP-1. Cardiovasc Res 47:49–56

41. Czibik G, Wu Z, Berne G, Tarkka M, Vaage J, Laurikka J, Jarvinen O, Valen G (2008) Human adaptation to ischemia by preconditioning or unstable angina: involvement of nuclear factor kappa B, but not hypoxia-inducible factor 1 alpha in the heart. Eur J Cardiothorac Surg 34:976–984. https ://doi. org/10.1016/j.ejcts .2008.07.066

42. Tahepold P, Vaage J, Starkopt J, Valen G (2003) Hyperoxia elicits myocardial protection through a nuclear factor kappa B-dependent mechanism in the rat heart. J Thorac Cardiovasc Surg 125:650–660. https ://doi.org/10.1067/mtc.2003.36 43. Xuan YT, Tang XL, Banerjee S, Takano H, Li RC, Han H,

Qlu Y, Li JJ, Bolli R (1999) Nuclear factor kappa B plays an essential role in the late phase of ischemic preconditioning in conscious rabbits. Circ Res 84:1095–1109

44. Misra A, Haudek SB, Knuefermann P, Vallejo JG, Chen ZJ, Michael LH, Sivasubramanian N, Olson EN, Entman ML, Mann DI (2003) Nuclear factor kappa B protects the adult cardiac myocyte against ischemia-induced apoptosis in a murine model of acute myocardial infarction. Circulation 108:3075–3078. https ://doi.org/10.1161/01.CIR.00001 08929 .93074 .0B 45. Libby P (2013) Mechanisms of acute coronary syndromes and

their implications for therapy. N Engl J Med 368:2004–2013. https ://doi.org/10.1056/NEJMr a1216 063

46. Buffon A, Basucci LM, Liuzzo G (2002) Widespread coronary inflammation in unstable angina. N Engl J Med 347:5–12. https ://doi.org/10.1056/NEJMo a0122 95

47. Bogaty P, Boyer L, Simard S, Dauwe F, Dupuis R, Verret B, Huynh T, Betrand F, Dagenais GR, Brophy JM (2008) The RISCA (Recurrence and Inflammation in the Acute Coronary Syndromes) Study. Clinical utility of C-reactive protein meas-ured ad admission, hospital discharge and 1 month later to predict outcome in patients with acute coronary syndrome. J Am Coll Cardiol 51(24):2339–2346. https ://doi.org/10.1016/j. jacc.2008.03.019

48. Liuzzo G, Biasucci LM, Gallimore JR (1994) The prognos-tic value of C-reactive protein and serum amyloid A protein in severe unstable angina. N Eng J Med 331:417–424. https ://doi. org/10.1056/NEJM1 99408 18331 0701

49. Biasucci LM, Liuzzo G, Grillo RL (1999) Elevated levels of C-reactive protein at discharge in patients with unstable angina predict recurrent instability. Circulation 99:855–860

50. Liuzzo G, Santamaria M, Biasucci LM, Narducci M, Colafranc-esco V, Porto A, Brugaletta S, Pinnelli M, Rizzello V, Maseri A, Crea F (2007) Persistent activation of Nuclear Factor Kappa-B signaling pathway in patients with unstable angina and elevated levels of C-reactive protein. J Am Coll Cardiol 49:185–194. https ://doi.org/10.1016/j.jacc.2006.07.071

51. Lapponi MJ, Carestia A, Landoni VI, Rivadeneyra L, Etulain J, Negrotto S, Pozner RG, Schattner M (2013) Regulation of neu-trophil extracellular trap formation by anti-inflammatory drugs. J Pharmacol Exp Ther 345(3):430–437. https ://doi.org/10.1124/ jpet.112.20287 9

52. Yin MJ, Yamamoto Y, Gaynor RB (1998) The anti-inflammatory agent aspirin and salicylate inhibit the activity of IkB kinase beta. Nature 396:77–80. https ://doi.org/10.1038/23948

53. Mc Donald PP, Bald A, Cassatella MA (1997) Activation of the NF-kappaB pathway by inflammatory stimuli in human neutro-phils. Blood 89:3421–3433

(9)

54. Massberg S, Grahl L, von Bruehl ML (2010) Reciprocal coupling of coagulation and innate immunity via neutrophil serine pro-teases. Nat Med 16:887–896. https ://doi.org/10.1038/nm.2184 55. Ross R (1999) Atherosclerosis: an inflammatory disease. N Engl

J Med 340:115–126

56. Ridker PM, Rifai N, Stampfer MJ, Hennekens CH (2000) Plasma concentration of interleukin-6 and the risk of future myocar-dial infarction among apparently healthy men. Circulation 101:1767–1772

57. Libby P, Ridker PM, Hansson GK (2009) Inflammation in athero-sclerosis: from pathophysiology to practice. J Am Coll Cardiol 54:2129–2138. https ://doi.org/10.1016/j.jacc.2009.09.009 58. Biasucci LM, Vitelli A, Liuzzo G, Altamura S, Caligiuri G,

Monaco C, Rebuzzi AG, Ciliberto G, Maseri A (1996) Elevated levels of Interleukin-6 in unstable angina. Circulation 94:874–877 59. Dinarello CA (2000) The role of the interleukin-1-receptor antag-onist in blocking inflammation mediated by interleukin-1. N Engl J Med 343:732–734. https ://doi.org/10.1056/NEJM2 00009 07343 1011

60. Dinarello CA (2009) Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol 27:519–550. https ://doi.org/10.1146/annur ev.immun ol.02190 8.13261 2

61. Sims JE, Smith DE (2010) The IL-1 family: regulators of immu-nity. Nat Rev Immunol 10:89–102. https ://doi.org/10.1038/nri26 91

62. Stutz A, Golenbock DT, Latz E (2009) Inflammasomes: too big to miss. J Clin Invest 119:3502–3511. https ://doi.org/10.1172/JCI40 599

63. Ogura Y, Sutterwala FS, Flavell RA (2006) The inflammasome: first line of the immune response to cell stress. Cell 126:659–662. https ://doi.org/10.1016/j.cell.2006.08.002

64. Rajama K, Lappalainen J, Oorni K (2010) Cholesterol crystals activate the NLRP3 inflammasome in human macrophages: a novel link between cholesterol metabolism and inflammation. PLoS ONE 5:e11765. https ://doi.org/10.1371/journ al.pone.00117 65

65. Ridker P, Howard C, Walter V, Everett B, Libby P, Hensen J, Thuren T (2012) Effects of interleukin-1 inhibition with canaki-numab on hemoglobin A1c, lipids, C-reactive protein, interleu-kin-6, and fibrinogen: a phase IIb randomized, placebo-controlled trial. Circulation 126:2739–2748. https ://doi.org/10.1161/CIRCU LATIO NAHA.112.12255 6

66. Warnatsch A, Ioannou M, Wang Q, Papayannopoulos V (2015) Neutrophil extracellular traps license macrophages for cytokine production in atherosclerosis. Science 349(6245):316–320. https ://doi.org/10.1126/scien ce.aaa80 64

67. Joshi MB, Lad A, Prasad AB, Balakrishnan A, Ramachandra L, Satyamoorthy K (2013) High glucose modulates IL-6 medi-ated immune homeostasis through impeding neutrophil extra-cellular trap formation. FEBS Lett 587:2241–2246. https ://doi. org/10.1016/j.febsl et.2013.05.053

68. Ikeda U, Ito T, Shimada K (2001) Interleukin-6 and acute coro-nary syndrome. Clin Cardiol 24(11):701–704

69. Rus HG, Vlaicu R, Niculescu F (1996) Interleukin-6 and interleu-kin-8 protein and gene expression in human arterial atheroscle-rotic wall. Atherosclerosis 127(2):263–271

70. Manten A, de Winter RJ, Minnema MC, ten Cate H, Lijmer JG, Adams R, Peters RJ, van Deventer SJ (1998) Procoagulant and proinflammatory activity in acute coronary syndromes. Cardio-vasc Res 40(2):389–395

71. Wang XH, Liu SQ, Wang YL, Jin Y (2014) Correlation of serum high-sensitivity C-reactive protein and interleukin-6 in patients with acute coronary syndrome. Genet Mol Res 13(2):4260–4266 72. Lai CL, Ji YR, Liu XH, Xing JP, Zhao JQ (2011) Relationship

between coronary atherosclerosis plaque characteristics and high sensitivity C-reactive proteins, interleukin-6. Chin Med J (Eng) 124(16):2452–2456

73. Van Lenten BJ, Hama SY, de Beer FC, Stafforini DM, McIntyre TM, Prescott SM, La Du BN, Fogelman AM, Navab M (1995) Anti-inflammatory HDL becomes pro-inflammatory during the acute phase response. Loss of protective effect of HDL against LDL oxidation in aortic wall cell cocultures. J Clin Investig 96(6):2758–2767. https ://doi.org/10.1172/JCI11 8345

74. Neumann FJ, Ott I, Marx N, Luther T, Kenngott S, Gawaz M, Kotzsch M, Schömig A (1997) Effect of human recombinant interleukin-6 and interleukin-8 on monocyte procoagulant activ-ity. Arterioscler Thromb Vasc Biol 17(12):3399–3405

75. Gwechenberger M, Mendoza LH, Youker KA, Frangogiannis NG, Smith CW, Michael LH, Entman ML (1999) Cardiac myocytes produce interleukin-6 in culture and in viable border zone of rep-erfused infarctions. Circulation 99(4):546–551

76. Caselli C, De Graaf MA, Lorenzoni V, Rovai D, Marinelli M, Del Ry S, Giannessi D, Bax JJ, Neglia D, Schlte AJ (2015) HDL cholesterol, leptin and interleukin-6 predict high risk coronary anatomy assessed by CT angiography in patients with stable chest pain. Atherosclerosis 241(1):55–61. https ://doi.org/10.1016/j. ather oscle rosis .2015.04.811

77. Koyama K, Yoneyama K, Mitarai T, Ishibashi Y, Takahashi E, Kongoji K, Harada T, Akashi YJ (2015) Association between inflammatory biomarkers and thin-cap fibroatheroma detected by optical coherence tomography in patients with coronary heart disease. Arch Med Sci 11(3):505–512. https ://doi.org/10.5114/ aoms.2015.52352

78. Plutzki J (2001) Inflammatory pathways in atherosclerosis and acute coronary syndromes. Am J Cardiol 88(8A):10K–15K 79. Brasier AR (2010) The nuclear factor-kB—interleukin-6

signal-ling pathway mediating vascular inflammation. Cardiovasc Res 86:211–218. https ://doi.org/10.1093/cvr/cvq07 6

80. Libermann TA, Baltimore D (1990) Activation of interleukin-6 gene expression through the NF-kappa B transcription factor. Mol Cell Biol 10(5):2327–2334

81. Lippi G, Sanchis-Gomar F, Cervellin G (2015) Cell-free DNA for diagnosing myocardial infarction: not ready for prime time. Clin Chem Lab Med 53(12):1895–1901. https ://doi.org/10.1515/ cclm-2015-0252

82. Tonello S, Rizzia M, Migliario M, Rocchetti V, Renò F (2017) Low concentrations of neutrophil extracellular traps induce prolif-eration in human keratinocytes via NF-kB activation. J Dermatol Sci 88:110–116

Riferimenti

Documenti correlati

Conversely, AR following challenge with the calcium ionophore A23187 was less significantly correlated with the percentage fertilization rate (P &lt; 0.05), and showed lower

scontro di classe 150. “Il dibattito politico e le decisioni assunte hanno riguardato solo gli strati superiori dell’organizzazione, svuotando i Circoli di

6.13 Trend of stopping distance evaluated with trigger method re- spect to tire wear and asphalt temperature, for the fourth test

Looking at the motif family organization allows to tackle the evolution of transcriptional regulation and identify global trends in comparative genomics, since it does not require

In addition, the LA group showed RFRs to fearful and happy faces in congruent muscles at about 625 ms after stimulus presentation; the peak of activation at 625 ms in the

I Musei visitabili sono: Museo di Paleontologia (Sala dei Dinosauri) e Museo di Zoologia e Anatomia Comparata, in via Università 4; Gemma 1786 (minerali, rocce, carte), in

☞ Indipendentemente dalla causa, lo shock è una ipo-perfusione sistemica dovuta a riduzione della gittata cardiaca o del volume di sangue efettivamente circolante. Il risultato fnale

However, since the γ-ray spectrum for Period D is much softer than the exceptionally hard γ-ray spectrum for Period B, in model D1 we need to adopt a break in the electron